Top Banner
1 Molecular mechanism of microtubule nucleation from gamma-tubulin ring complex 1 2 Akanksha Thawani 1 , Howard A Stone 2 , Joshua W Shaevitz 3,4 , Sabine Petry 5,* 3 4 1 Department of Chemical and Biological Engineering, Princeton University 5 2 Department of Mechanical and Aerospace Engineering, Princeton University 6 3 Lewis-Sigler Institute for Integrative Genomics, Princeton University 7 4 Department of Physics, Princeton University 8 5 Department of Molecular Biology, Princeton University, United States 9 10 * Correspondence to: Sabine Petry ([email protected]) 11 not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which was this version posted November 23, 2019. ; https://doi.org/10.1101/853010 doi: bioRxiv preprint
51

Molecular mechanism of microtubule nucleation from gamma ...61 TuRC (Fig. S1E), we showed that γ-TuRC caps the MT minus-end, while only the plus-end 62 polymerizes. Altogether, our

Jan 27, 2021

Download

Documents

dariahiddleston
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
  • 1

    Molecular mechanism of microtubule nucleation from gamma-tubulin ring complex 1

    2

    Akanksha Thawani1, Howard A Stone2, Joshua W Shaevitz3,4, Sabine Petry5,* 3

    4

    1Department of Chemical and Biological Engineering, Princeton University 5

    2Department of Mechanical and Aerospace Engineering, Princeton University 6

    3Lewis-Sigler Institute for Integrative Genomics, Princeton University 7

    4Department of Physics, Princeton University 8

    5Department of Molecular Biology, Princeton University, United States 9

    10

    * Correspondence to: Sabine Petry ([email protected]) 11

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 2

    Abstract 12

    Determining how microtubules (MTs) are nucleated is essential for understanding how the 13

    cytoskeleton assembles. Yet, half a century after the discovery of MTs and ab-tubulin subunits 14

    and decades after the identification of the γ-tubulin ring complex (γ-TuRC) as the universal MT 15

    nucleator, the underlying mechanism largely remains a mystery. Using single molecule studies, 16

    we uncover that γ-TuRC nucleates a MT more efficiently than spontaneous assembly. The laterally 17

    interacting array of γ-tubulins on γ-TuRC facilitates the lateral association of αβ-tubulins, while 18

    longitudinal affinity between γ/αβ-tubulin is surprisingly weak. During nucleation, 3-4 αβ-tubulin 19

    dimers bind stochastically to γ-TuRC on average until two of them create a lateral contact and 20

    overcome the nucleation barrier. Although γ-TuRC defines the nucleus, XMAP215 significantly 21

    increases reaction efficiency by facilitating ab-tubulin incorporation. In sum, we elucidate how 22

    MT initiation occurs from γ-TuRC and determine how it is regulated. 23

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 3

    Introduction 24

    Microtubules (MTs) enable cell division, motility, intracellular organization and transport. MTs 25

    were found to consist of αβ-tubulin dimers fifty years ago, yet, how MTs are nucleated in the cell 26

    to build the cytoskeleton remains poorly understood1–3. 27

    MTs assemble spontaneously from αβ-tubulin subunits in vitro via the cooperative assembly 28

    of many tubulin dimers and hence this process displays a kinetic barrier4–8. Consequently, 29

    spontaneous MT nucleation is rarely observed in cells9,10. Instead, the major MT nucleator γ-30

    tubulin is required in vivo9–11. γ-tubulin forms a 2.2 megadalton, ring-shaped complex with γ-31

    tubulin complex proteins (GCPs), known as the γ-Tubulin Ring Complex (γ-TuRC) 12–16. γ-TuRC 32

    has been proposed to template the assembly of αβ-tubulin dimers into a ring, resulting in nucleation 33

    of a MT15–21. However, kinetic measurements that provide direct evidence for this hypothesis have 34

    been lacking and several important questions about how γ-TuRC nucleates MTs have remained 35

    unanswered. 36

    In the absence of purified g-TuRC and an assay to visualize MT nucleation events from 37

    single g-TuRC molecules in real time, recent studies used alternative MT assembly sources, such 38

    as spontaneous MT assembly or stabilized MTs with blunt ends hypothesized to resemble the γ-39

    TuRC interface. Based on these alternatives, competition between growth and catastrophe of the 40

    nascent plus-end was proposed to yield the nucleation barrier in the cell22,23, but this has not been 41

    examined with the nucleator g-TuRC. Recently, the MT polymerase XMAP215 was identified as 42

    an essential MT nucleation factor in vivo, which synergistically nucleates MTs with γ-TuRC24–26. 43

    Yet, the specific roles of XMAP215 and γ-TuRC in MT nucleation have yet to be discovered. 44

    To explore the mechanism of MT nucleation, we reconstituted and visualized MT nucleation 45

    by γ-TuRC live with single molecule resolution. We uncover the molecular composition of the 46

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 4

    MT nucleus, and determine the roles XMAP215 and γ-TuRC in MT nucleation. 47

    48

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 5

    Results 49

    50

    Reconstituting and visualizing microtubule nucleation from γ-TuRC 51

    To study how γ-TuRC nucleates MTs (Fig. 1A), we purified endogenous γ-TuRC from Xenopus 52

    egg extracts and biotinylated the complexes to immobilize them on functionalized glass (Fig. S1A-53

    C). Upon perfusing fluorescent αβ-tubulin, we visualized MT nucleation live with total internal 54

    reflection fluorescence microscopy (TIRFM). Strikingly, MT nucleation events occurred 55

    specifically from single γ-TuRC molecules (Fig. 1B; Fig. S1D and Movie S1-2). Kymographs 56

    revealed that attached γ-TuRC assembled ab-tubulin into a MT de novo starting from zero length 57

    within the diffraction limit of light microscopy (Fig. 1C), ruling out an alternative model where 58

    MTs first spontaneously nucleate and then become stabilized via γ-TuRC. By observing the 59

    fiduciary marks on the MT lattice (Fig. 1C) and generating polarity-marked MTs from attached g-60

    TuRC (Fig. S1E), we showed that γ-TuRC caps the MT minus-end, while only the plus-end 61

    polymerizes. Altogether, our results show that γ-TuRC directly nucleates MTs. 62

    63

    Defining the microtubule nucleus on γ-TuRC 64

    To determine how γ-TuRC nucleates MTs, we measured the kinetics of MT nucleation for a 65

    constant density of γ-TuRC and increasing αβ-tubulin concentration (Fig. 1D and Movie S3). 66

    Surprisingly, γ-TuRC nucleated MTs starting from 7 µM tubulin (Fig. 1D), which is higher than 67

    the minimum tubulin concentration (C*) needed for growth at pre-formed MT plus-ends (C* = 1.4 68

    µM, Fig. 1E). Furthermore, the number of MTs nucleated from γ-TuRC increased non-linearly 69

    with tubulin concentration as opposed to the linear increase in MT’s growth-speed with tubulin 70

    concentration (Fig. 1E). By measuring the number of MTs nucleated over time with varying ab-71

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 6

    tubulin concentration (Fig. 1F), we calculated the rate of MT nucleation. The power-law 72

    dependence on tubulin concentration (Fig. 1G) yields the number of ab-tubulin dimers, 3.7 ± 0.5, 73

    that initiate MT assembly from g-TuRC (Fig. 1G). Thus, the cooperative assembly of 3-4 tubulin 74

    subunits on g-TuRC represents the most critical, rate-limiting step in MT nucleation. 75

    76

    Efficiency of γ-TuRC-mediated nucleation 77

    Based on the traditional, fixed, end-point assays for MT nucleation with large error margins, g-78

    TuRC was believed to be a poor nucleator14. To measure the efficiency of γ-TuRC-mediated MT 79

    nucleation, we compared it with spontaneous MT nucleation in our live TIRFM assay (Fig. 1H). 80

    In contrast to γ-TuRC-mediated nucleation, a high concentration of 14 µM tubulin was required 81

    for spontaneous assembly of MTs, after which both the plus- and minus-ends polymerize (Fig. 1H, 82

    Fig. S1F and Movie S4). The number of MTs assembled as a function of the ab-tubulin 83

    concentration displayed a power-law dependence with the exponent of 8 ± 1 (Fig. 1I), indicating 84

    a highly cooperative process that requires 8 ab-tubulin dimers in a rate-limiting intermediate, in 85

    agreement with previous reports (Fig. 1H schematic, refs 4,8). In conclusion, γ-TuRC nucleates 86

    MTs significantly more efficiently (Fig. S1G), because its critical nucleus requires less than half 87

    the number of ab-tubulin dimers compared to spontaneous assembly. 88

    89

    Does γ-TuRC nucleate a microtubule via the blunt plus-end model? 90

    It has been widely proposed that the g-tubulin ring on γ-TuRC resembles the blunt plus-end of a 91

    MT formed by a ring of ab-tubulins20,22,27. To test this proposition, we generated stabilized MT 92

    seeds with blunt ends as described recently22 and observed MT assembly from αβ-tubulin dimers 93

    (Fig. 2A). At a minimum concentration of 2.45 µM, approaching the critical concentration needed 94

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 7

    for polymerization, a large proportion of pre-formed MT seeds assembled MTs immediately (Fig. 95

    S2A-B, Fig. 2A and Movie S5). The measured reaction kinetics (Fig. 2B) as a function of the ab-96

    tubulin concentration was used to obtain a power-law of the nucleation rate, 1.2 ± 0.4 (Fig. 2C). 97

    This demonstrates that blunt MT seeds assemble tubulin dimers into a lattice in a non-cooperative 98

    manner, where a single ab-tubulin dimer suffices to overcome the rate-limiting step resembling 99

    the polymerization of a MT. Thus, the kinetics of γ-TuRC-mediated MT nucleation does not 100

    resemble a blunt MT plus-end. 101

    102

    Molecular insight into microtubule nucleation by g-TuRC 103

    We hypothesized that γ-tubulin’s binding properties with ab-tubulin at the nucleation interface γ-104

    TuRC could provide insight into the mechanism of nucleation. We purified γ-tubulin, which 105

    assembles into higher order oligomers in physiological buffer 24 and strikingly, into filaments at 106

    high g-tubulin concentrations (Fig. S2C). Because γ-tubulins have been shown to arrange laterally, 107

    as observed previously in its crystallized form28, a plus-ends outward orientation of g-tubulin 108

    molecules could form a nucleation interface. 109

    Surprisingly, the γ-tubulin oligomers efficiently nucleated MTs from αβ-tubulin subunits 110

    (Fig. 2D and Movie S6) and even more strikingly, capped MT minus-ends while allowing newly 111

    generated MT plus-ends to polymerize (Fig. 2E). This activity is similar to that of γ-TuRC, 112

    suggesting that lateral γ-tubulin arrays on the nucleation interface of γ-TuRC are sufficient to 113

    nucleate MTs. 114

    Knowing that lateral γ-tubulin arrays in purified γ-tubulin oligomers and within g-TuRC 115

    nucleate MTs, we hypothesized that the longitudinal affinity between γ-tubulin and αβ-tubulin at 116

    the interface of γ-TuRC could be critical in regulating its nucleation efficiency. Using biolayer 117

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 8

    interferometry, we compared the interaction of ab-tubulin dimers with themselves versus with g-118

    tubulin. Specific interactions between probe-bound αβ-tubulin and increasing concentrations of 119

    unlabeled αβ-tubulin were measured (Fig. 2F), which must be longitudinal based on the observed 120

    protofilaments in the ab-tubulin sample by EM (Fig. S2D). In contrast, no significant binding 121

    between monomeric γ-tubulin and αβ-tubulin was detected (Fig. 2F), suggesting that the 122

    heterogenous longitudinal affinity between g-tubulin and ab-tubulin on the nucleation interface 123

    may be weaker compared to αβ-tubulin with another ab-tubulin molecule that occurs when the 124

    MT lattice polymerizes. In sum, the difference in interaction strength is the basis for the kinetic 125

    barrier we observed with g-TuRC but not with a blunt MT plus-end, which we summarize with an 126

    interface interaction model (Fig. 2G). 127

    We next asked how 3-4 tubulin dimers formed the rate-limiting species during γ-TuRC 128

    nucleation. In stochastic simulations, the 13 available binding sites on g-tubulin molecules within 129

    γ-TuRC were allowed to be occupied at random with αβ-tubulin subunits. We then assessed how 130

    many ab-tubulin dimers need to assemble on g-TuRC to obtain two αβ-tubulin molecules on 131

    neighboring sites and form a favorable configuration with a lateral contact between the two αβ-132

    tubulins (Fig. 2H). The simulations show that 3.7 ± 1 tubulin dimers assemble on γ-TuRC to form 133

    the first lateral contact between two αβ-tubulins (Fig. 2H), in striking agreement with the critical 134

    nucleus size we measured. In sum, our data shows that a lateral γ-tubulin array positioned by γ-135

    TuRC promotes MT nucleation. The low g-tubulin:ab-tubulin affinity requires binding of 3-4 αβ-136

    tubulin dimers to g-TuRC to form the first lateral contact between two αβ-tubulin dimers and 137

    overcome the kinetic barrier before entering the MT polymerization phase. This nucleation barrier, 138

    in turn, provides the ability to further modulate MT nucleation via other factors. 139

    140

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 9

    Regulation of γ-TuRC mediated nucleation by microtubule associated proteins 141

    Recent work suggested that MT-associated proteins (MAPs), which stabilize or destabilize MT 142

    plus-ends, influence MT nucleation in an analogous fashion7,22,23,27. We assessed this hypothesis 143

    for MT nucleation by γ-TuRC. The protein TPX2 functions as an anti-catastrophe factor in vitro 144

    22,23 and has been suggested to directly stimulate γ-TuRC-mediated nucleation21,29–31. Strikingly, 145

    although TPX2 binds along the MT lattice, it does not increase nucleation activity of γ-TuRC (Fig. 146

    3A and Movie S7). Similarly, the catastrophe factor EB1 does not decrease the nucleation activity 147

    of γ-TuRC (Fig. S3A and Movie S8). Thus, in agreement with our previous results (Figs. 1 and 148

    2A-B), destabilization of MT plus-ends and a competition between 149

    polymerization/depolymerization is not sufficient to explain the properties of MT nucleation from 150

    g-TuRC. Not surprisingly, decreasing the net rate of incorporation of tubulin into a MT using 151

    Stathmin, which sequesters tubulin dimers32,33, or MCAK, which removes tubulin dimers from the 152

    MT lattice and prevents polymerization34,35, decreased the number of MTs generated from γ-TuRC 153

    (Fig. S3B). 154

    155

    How do γ-TuRC and XMAP215 synergistically nucleate microtubules? 156

    At low tubulin concentration of 3.5 µM and 7 µM, where either none or very little MT nucleation 157

    occurs from γ-TuRCs alone respectively, the addition of XMAP215 induced many surface-158

    attached γ-TuRCs to nucleate MTs resulting in significant increase in MT nucleation rate (Fig. 3B-159

    C and Movie S9). XMAP215 effectively decreases the minimal tubulin concentration necessary 160

    for MT nucleation from γ-TuRC to 1.6 µM, which is very close to that needed for plus-end 161

    polymerization. What is the sequence of events that leads to synergistic MT nucleation? By 162

    directly visualizing γ-TuRC and XMAP215 molecules during the nucleation reaction, we found 163

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 10

    that XMAP215 and γ-TuRC molecules first formed a complex from which a MT was nucleated 164

    (Fig. 3D and Movie S11). For 76% of the events (n=56), XMAP215 visibly persisted between 3 165

    to over 300 seconds on γ-TuRC before MT nucleation, and with a 50% probability XMAP215 166

    remained on the minus-end together with γ-TuRC (n=58). 167

    Could XMAP215 accelerate nucleation by altering the critical tubulin nucleus that 168

    assembles during γ-TuRC-mediated nucleation? Titrating tubulin at constant γ-TuRC and 169

    XMAP215 concentrations (Fig. S4A and Movies S10) yielded a similar power-law dependence 170

    between the MT nucleation rate and tubulin concentration (Fig. 3E). The resulting critical nucleus 171

    size of 3.2 ± 1.2 is very similar to that for γ-TuRC alone (Fig. 3E). Moreover, the C-terminus of 172

    XMAP215 (TOG5 and C-terminal domain), which directly interacts with γ-tubulin but not with 173

    αβ-tubulin24, does not enhance MT nucleation from γ-TuRC (Fig. S4B). Altogether, γ-TuRC 174

    determines the critical nucleus of ab-tubulin dimers for MT nucleation (Fig. 2H). XMAP215, 175

    which directly binds to g-tubulin via its C-terminal domain, does not appear to activate g-TuRC 176

    via a conformational change, but likely relies on N-terminal TOG domains to increase ab-tubulin 177

    incorporation by effectively increasing the local ab-tubulin concentration, and thereby promoting 178

    MT nucleation. 179

    180

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 11

    Discussion 181

    Decades after the discovery of ab-tubulin and MTs and the identification of γ-TuRC as the 182

    universal MT nucleator17–19, it has remained poorly understood how MTs are being nucleated7,20,21. 183

    Here, we show that γ-TuRC-mediated MT nucleation is more efficient than spontaneous MT 184

    assembly, requiring fewer tubulin dimers to form the rate-limiting reaction intermediate. This 185

    explains why MTs do not form spontaneously in the cell and why γ-TuRC is essential, addressing 186

    a long debate on g-TuRC’s MT nucleation activity and requirement36–38. Spontaneous MT 187

    assembly requires higher tubulin concentrations and occurs due to stronger longitudinally-188

    interacting αβ/αβ-tubulin and weaker lateral interactions. In contrast, g-TuRC-mediated 189

    nucleation, driven by the lateral adjacency of the g-tubulins on the nucleation interface, is sufficient 190

    to overcome the intrinsically very weak ab-tubulin lateral interaction, thereby potentiating MT 191

    nucleation. Thus, we propose that, in metazoans analogous to the S. cerevisiae γ-TuSC rings15,16, 192

    GCPs within γ-TuRC restrict the number of laterally-arranged g-tubulin subunits, and position 193

    them in the right geometry to template 13-pf MTs. Finally, our results show that 3-4 ab-tubulin 194

    form the critical nucleus on g-TuRC, not 1 or 13 which would have been expected from previous 195

    mechanistic hypotheses20. We find that on average 3-4 ab-tubulin dimers assemble on g-TuRC to 196

    form the first lateral ab-/ab-tubulin contact and overcome the kinetic barrier that results from low 197

    longitudinal affinity between g-:ab-tubulin on g-TuRC. However, alternative reaction 198

    intermediates during nucleation from g-TuRC may exist. In the future, it will be important to 199

    visualize the nucleation intermediates on g-TuRC, develop molecular simulations with 200

    experimentally derived affinities at various interaction interfaces and evaluate whether additional 201

    effects from tubulin straightening play a significant role in MT nucleation in the cell. 202

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 12

    The intermediate level of MT nucleation efficiency afforded by g-TuRC allows other 203

    factors to further modulate its efficiency. As such, XMAP215 accelerates MT nucleation from γ-204

    TuRC, while not altering the geometry of the ab-tubulin nucleus on g-TuRC or directly activating 205

    g-TuRC. Future studies will be necessary to define the modes by which XMAP215 contributes to 206

    g-TuRC-mediated MT nucleation, such as increasing the probability of the g/ab-tubulin interaction 207

    or promoting straightening of incoming tubulin dimers. Our findings suggest that influencing g/ab-208

    tubulin interaction favorably or unfavorably may underlie a dominant mechanism for regulating 209

    nucleation in the cell by other, yet unidentified nucleation factors. Additionally, g-TuRC’s activity 210

    is further regulated via accessory proteins such as CDK5RAP2, and NME72,20,39,40. While the 211

    mechanisms of these additional regulation layers are yet to be defined, the insights on MT 212

    nucleation by γ-TuRC and XMAP215 provide an essential basis to build upon. Finally, this work 213

    opens the door to reconstitute cellular structures in vitro using MT nucleation from γ-214

    TuRC/XMAP215 to further our understanding of how the cytoskeleton is generated to support cell 215

    function. 216

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 13

    Supplementary Information 217

    Supplementary Information includes four figures and ten videos. 218

    219

    Acknowledgements 220

    We thank David Agard, Tim Mitchison, Michelle Moritz and Petry lab members for discussions. 221

    This work was supported by an American Heart Association predoctoral fellowship 222

    17PRE33660328 and a Princeton University Honorific Fellowship (both to AT), the NIH New 223

    Innovator Award 1DP2GM123493, Pew Scholars Program in the Biomedical Sciences 00027340, 224

    David and Lucile Packard Foundation 2014-40376 (all to SP), and the Center for the Physics of 225

    Biological Function sponsored by the National Science Foundation grant PHY-1734030. 226

    227

    Author contributions 228

    A.T. designed and performed research, analyzed the data and wrote the manuscript. S.P., J.W.S. 229

    and H.A.S. supervised research and wrote the manuscript. 230

    231

    Competing financial interests 232

    The authors declare no competing financial interests. 233

    234

    Abbreviations List 235

    Microtubule (MT) 236

    Microtubule associated protein (MAP) 237

    Gamma-tubulin (γ-tubulin) and Gamma-tubulin ring complex (γ-TuRC) 238

    Gamma-tubulin complex protein (GCP) 239

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 14

    Protofilament (pf) 240

    Electron microscopy (EM) 241

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 15

    References 242

    1. Brugués, J., Nuzzo, V., Mazur, E. & Needleman, D. J. Nucleation and transport organize 243

    microtubules in metaphase spindles. Cell 149, 554–564 (2012). 244

    2. Petry, S. & Vale, R. D. Microtubule nucleation at the centrosome and beyond. Nat. Cell Biol. 245

    17, 1089–1093 (2015). 246

    3. Wu, J. & Akhmanova, A. Microtubule-Organizing Centers. Annu. Rev. Cell Dev. Biol. 33, 51–247

    75 (2017). 248

    4. Voter, W. A. & Erickson, H. P. The kinetics of microtubule assembly. Evidence for a two-249

    stage nucleation mechanism. J. Biol. Chem. 259, 10430–10438 (1984). 250

    5. Portran, D., Schaedel, L., Xu, Z., Théry, M. & Nachury, M. V. Tubulin acetylation protects 251

    long-lived microtubules against mechanical ageing. Nat. Cell Biol. 19, 391–398 (2017). 252

    6. Rice, L., Moritz, M. & Agard, D. A. Microtubules form by progressively faster tubulin 253

    accretion, not by nucleation-elongation. http://biorxiv.org/lookup/doi/10.1101/545236 254

    (2019) doi:10.1101/545236. 255

    7. Roostalu, J. & Surrey, T. Microtubule nucleation: beyond the template. Nat. Rev. Mol. Cell 256

    Biol. 18, 702–710 (2017). 257

    8. Flyvbjerg, H., Jobs, E. & Leibler, S. Kinetics of self-assembling microtubules: an ‘inverse 258

    problem’ in biochemistry. Proc. Natl. Acad. Sci. U.S.A. 93, 5975–5979 (1996). 259

    9. Hannak, E. et al. The kinetically dominant assembly pathway for centrosomal asters in 260

    Caenorhabditis elegans is gamma-tubulin dependent. J. Cell Biol. 157, 591–602 (2002). 261

    10. Groen, A. C., Maresca, T. J., Gatlin, J. C., Salmon, E. D. & Mitchison, T. J. Functional 262

    overlap of microtubule assembly factors in chromatin-promoted spindle assembly. Mol. Biol. 263

    Cell 20, 2766–2773 (2009). 264

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 16

    11. Oakley, C. E. & Oakley, B. R. Identification of gamma-tubulin, a new member of the tubulin 265

    superfamily encoded by mipA gene of Aspergillus nidulans. Nature 338, 662–664 (1989). 266

    12. Moritz, M., Braunfeld, M. B., Sedat, J. W., Alberts, B. & Agard, D. A. Microtubule 267

    nucleation by gamma-tubulin-containing rings in the centrosome. Nature 378, 638–640 268

    (1995). 269

    13. Moritz, M., Zheng, Y., Alberts, B. M. & Oegema, K. Recruitment of the gamma-tubulin ring 270

    complex to Drosophila salt-stripped centrosome scaffolds. J. Cell Biol. 142, 775–786 (1998). 271

    14. Zheng, Y., Wong, M. L., Alberts, B. & Mitchison, T. Nucleation of microtubule assembly by 272

    a gamma-tubulin-containing ring complex. Nature 378, 578–583 (1995). 273

    15. Kollman, J. M., Polka, J. K., Zelter, A., Davis, T. N. & Agard, D. A. Microtubule nucleating 274

    gamma-TuSC assembles structures with 13-fold microtubule-like symmetry. Nature 466, 275

    879–882 (2010). 276

    16. Kollman, J. M. et al. Ring closure activates yeast γTuRC for species-specific microtubule 277

    nucleation. Nat. Struct. Mol. Biol. 22, 132–137 (2015). 278

    17. Moritz, M., Braunfeld, M. B., Guénebaut, V., Heuser, J. & Agard, D. A. Structure of the 279

    gamma-tubulin ring complex: a template for microtubule nucleation. Nat. Cell Biol. 2, 365–280

    370 (2000). 281

    18. Keating, T. J. & Borisy, G. G. Immunostructural evidence for the template mechanism of 282

    microtubule nucleation. Nat. Cell Biol. 2, 352–357 (2000). 283

    19. Wiese, C. & Zheng, Y. A new function for the gamma-tubulin ring complex as a microtubule 284

    minus-end cap. Nat. Cell Biol. 2, 358–364 (2000). 285

    20. Kollman, J. M., Merdes, A., Mourey, L. & Agard, D. A. Microtubule nucleation by γ-tubulin 286

    complexes. Nat. Rev. Mol. Cell Biol. 12, 709–721 (2011). 287

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 17

    21. Tovey, C. A. & Conduit, P. T. Microtubule nucleation by γ-tubulin complexes and beyond. 288

    Essays Biochem. 62, 765–780 (2018). 289

    22. Wieczorek, M., Bechstedt, S., Chaaban, S. & Brouhard, G. J. Microtubule-associated 290

    proteins control the kinetics of microtubule nucleation. Nat. Cell Biol. 17, 907–916 (2015). 291

    23. Roostalu, J., Cade, N. I. & Surrey, T. Complementary activities of TPX2 and chTOG 292

    constitute an efficient importin-regulated microtubule nucleation module. Nat. Cell Biol. 17, 293

    1422–1434 (2015). 294

    24. Thawani, A., Kadzik, R. S. & Petry, S. XMAP215 is a microtubule nucleation factor that 295

    functions synergistically with the γ-tubulin ring complex. Nat. Cell Biol. 20, 575–585 296

    (2018). 297

    25. Flor-Parra, I., Iglesias-Romero, A. B. & Chang, F. The XMAP215 Ortholog Alp14 Promotes 298

    Microtubule Nucleation in Fission Yeast. Curr. Biol. 28, 1681-1691.e4 (2018). 299

    26. Gunzelmann, J. et al. The microtubule polymerase Stu2 promotes oligomerization of the γ-300

    TuSC for cytoplasmic microtubule nucleation. Elife 7, (2018). 301

    27. Brouhard, G. J. & Rice, L. M. Microtubule dynamics: an interplay of biochemistry and 302

    mechanics. Nat. Rev. Mol. Cell Biol. 19, 451–463 (2018). 303

    28. Aldaz, H., Rice, L. M., Stearns, T. & Agard, D. A. Insights into microtubule nucleation from 304

    the crystal structure of human gamma-tubulin. Nature 435, 523–527 (2005). 305

    29. Alfaro-Aco, R., Thawani, A. & Petry, S. Structural analysis of the role of TPX2 in branching 306

    microtubule nucleation. J. Cell Biol. 216, 983–997 (2017). 307

    30. Zhang, R., Roostalu, J., Surrey, T. & Nogales, E. Structural insight into TPX2-stimulated 308

    microtubule assembly. Elife 6, (2017). 309

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 18

    31. Wiese, C. & Zheng, Y. Microtubule nucleation: gamma-tubulin and beyond. J. Cell. Sci. 310

    119, 4143–4153 (2006). 311

    32. Jourdain, L., Curmi, P., Sobel, A., Pantaloni, D. & Carlier, M. F. Stathmin: a tubulin-312

    sequestering protein which forms a ternary T2S complex with two tubulin molecules. 313

    Biochemistry 36, 10817–10821 (1997). 314

    33. Belmont, L. D. & Mitchison, T. J. Identification of a protein that interacts with tubulin 315

    dimers and increases the catastrophe rate of microtubules. Cell 84, 623–631 (1996). 316

    34. Howard, J. & Hyman, A. A. Microtubule polymerases and depolymerases. Curr. Opin. Cell 317

    Biol. 19, 31–35 (2007). 318

    35. Hunter, A. W. et al. The kinesin-related protein MCAK is a microtubule depolymerase that 319

    forms an ATP-hydrolyzing complex at microtubule ends. Mol. Cell 11, 445–457 (2003). 320

    36. Rogers, G. C., Rusan, N. M., Peifer, M. & Rogers, S. L. A multicomponent assembly 321

    pathway contributes to the formation of acentrosomal microtubule arrays in interphase 322

    Drosophila cells. Mol. Biol. Cell 19, 3163–3178 (2008). 323

    37. Raff, J. W. Phase Separation and the Centrosome: A Fait Accompli? Trends Cell Biol. 29, 324

    612–622 (2019). 325

    38. Woodruff, J. B. et al. The Centrosome Is a Selective Condensate that Nucleates Microtubules 326

    by Concentrating Tubulin. Cell 169, 1066-1077.e10 (2017). 327

    39. Choi, Y.-K., Liu, P., Sze, S. K., Dai, C. & Qi, R. Z. CDK5RAP2 stimulates microtubule 328

    nucleation by the gamma-tubulin ring complex. J. Cell Biol. 191, 1089–1095 (2010). 329

    40. Liu, P., Choi, Y.-K. & Qi, R. Z. NME7 is a functional component of the γ-tubulin ring 330

    complex. Mol. Biol. Cell 25, 2017–2025 (2014). 331

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 19

    41. Petry, S., Pugieux, C., Nédélec, F. J. & Vale, R. D. Augmin promotes meiotic spindle 332

    formation and bipolarity in Xenopus egg extracts. Proc. Natl. Acad. Sci. U.S.A. 108, 14473–333

    14478 (2011). 334

    42. Tan, S., Kern, R. C. & Selleck, W. The pST44 polycistronic expression system for producing 335

    protein complexes in Escherichia coli. Protein Expr. Purif. 40, 385–395 (2005). 336

    43. Ohi, R., Sapra, T., Howard, J. & Mitchison, T. J. Differentiation of cytoplasmic and meiotic 337

    spindle assembly MCAK functions by Aurora B-dependent phosphorylation. Mol. Biol. Cell 338

    15, 2895–2906 (2004). 339

    44. Reber, S. B. et al. XMAP215 activity sets spindle length by controlling the total mass of 340

    spindle microtubules. Nat. Cell Biol. 15, 1116–1122 (2013). 341

    45. Hannak, E. & Heald, R. Investigating mitotic spindle assembly and function in vitro using 342

    Xenopus laevis egg extracts. Nat Protoc 1, 2305–2314 (2006). 343

    46. Murray, A. W. & Kirschner, M. W. Cyclin synthesis drives the early embryonic cell cycle. 344

    Nature 339, 275–280 (1989). 345

    47. Bieling, P., Telley, I. A., Hentrich, C., Piehler, J. & Surrey, T. Fluorescence microscopy 346

    assays on chemically functionalized surfaces for quantitative imaging of microtubule, motor, 347

    and +TIP dynamics. Methods Cell Biol. 95, 555–580 (2010). 348

    349

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 20

    Methods 350

    351

    Purification of recombinant proteins 352

    C-terminal GFP was replaced with mCherry tag in the pET21a vector carrying EB141. Full-length 353

    TPX2 with N-terminal Strep II-6xHis-GFP-TEV site tags was cloned into pST50Tr-354

    STRHISNDHFR (pST50) vector42 using Gibson Assembly (New England Biolabs). N-terminal 355

    6xHis-tagged, Xenopus laevis Stathmin 1A was a gift from Christiane Wiese (University of 356

    Madison). N-terminal tagged 6xHis-TEV MCAK plasmid was a gift from Ryoma Ohi43. Wild-357

    type XMAP215 with C-terminal GFP-7xHis plasmid was a gift from Simone Reber44 and was used 358

    to clone XMAP215 with C-terminal SNAP-TEV-7xHis-StrepII tags, first into pST50 vector and 359

    further into pFastBac1 vector. TOG5-CT truncation of XMAP215 was produced by cloning amino 360

    acids 1091-2065 into pST50 vector with C-terminal GFP-7xHis-Strep tags. Human γ-tubulin TEV-361

    Strep II-6xHis tags was codon-optimized for Sf9 expression, synthesized (Genscript), and further 362

    cloned into pFastBac1 vector. 363

    EB1, TPX2, Stathmin and XMAP215 TOG5-CT used in this study were expressed in E. 364

    coli Rosetta2 cells (EMD Millipore) by inducing with 0.5-1 mM IPTG for 12-18 hours at 16°C or 365

    7 hours at 25°C. Wild-type XMAP215, MCAK and γ-tubulin were expressed and purified from 366

    Sf9 cells using Bac-to-Bac system (Invitrogen). The cells were lysed (EmulsiFlex, Avestin) and 367

    E. coli lysate was clarified by centrifugation at 13,000 rpm in Fiberlite F21-8 rotor (ThermoFisher) 368

    and Sf9 cell lysate at 50,000 rpm in Ti70 rotor (Beckman Coulter) for 30-45 minutes. 369

    EB1 and Stathmin were purified using His-affinity (His-Trap HP, GE Healthcare) by first 370

    binding in binding buffer (20mM NaPO4 pH 8.0, 500mM NaCl, 30mM Imidazole, 2.5mM PMSF, 371

    6mM BME) and eluting with 300mM Imidazole, followed by gel filtration (HiLoad 16/600 372

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 21

    Superdex, GE Healthcare) into CSF-XB buffer (100mM KCl, 10mM K-HEPES, 5mM K-EGTA, 373

    1mM MgCl2, 0.1mM CaCl2, pH 7.7 with 10% w/v sucrose). 374

    TPX2 was first affinity purified using Ni-NTA beads in binding buffer (50mM Tris-HCl 375

    pH 8.0, 750mM NaCl, 15mM Imidazole, 2.5mM PMSF, 6mM BME) and eluted with 200mM 376

    Imidazole. All protein was pooled and diluted 4-fold to 200mM final NaCl. Nucleotides were 377

    removed with a Heparin column (HiTrap Heparin HP, GE Healthcare) by binding protein in 378

    250mM NaCl and isocratic elution in 750mM NaCl, all solutions prepared in Heparin buffer 379

    (50mM Tris-HCl, pH 8.0, 2.5mM PMSF, 6mM BME). Peak fractions were pooled and loaded on 380

    to Superdex 200 pg 16/600, and gel filtration was performed in CSF-XB buffer. 381

    MCAK was first affinity purified by binding to His-Trap HP (GE Healthcare) in binding 382

    buffer (50mM NaPO4, 500mM NaCl, 6mM BME, 0.1mM MgATP, 10mM Imidazole, 1mM 383

    MgCl2, 2.5mM PMSF, 6mM BME, pH to 7.5), eluting with 300mM Imidazole, followed by gel-384

    filtration (Superdex 200 10/300 GL, GE Healthcare) in storage buffer (10 mM K-HEPES pH 7.7, 385

    300 mM KCl, 6mM BME, 0.1 mM MgATP, 1mM MgCl2, 10% w/v sucrose). 386

    XMAP215-GFP was purified using His-affinity (His-Trap, GE Healthcare) by binding in 387

    buffer (50mM NaPO4, 500mM NaCl, 20mM Imidazole, pH 8.0) and eluting in 500mM Imidazole. 388

    Peak fractions were pooled and diluted 5-fold with 50mM Na-MES pH 6.6, bound to a cation-389

    exchange column (Mono S 10/100 GL, GE Healthcare) with 50mM MES, 50mM NaCl, pH 6.6 390

    and eluted with a salt-gradient up to 1M NaCl. Peak fractions were pooled and dialyzed into CSF-391

    XB buffer. SNAP-tagged XMAP215 was first affinity purified with StrepTrap HP (GE Healthcare) 392

    with binding buffer (50mM NaPO4, 270mM NaCl, 2mM MgCl2, 2.5mM PMSF, 6mM BME, pH 393

    7.2), eluted with 2.5mM D-desthiobiotin, and cation-exchanged (Mono S 10/100 GL). Peak 394

    fractions were pooled, concentrated and reacted with 2-molar excess SNAP-substrate Alexa-488 395

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 22

    dye (S9129, NEB) overnight at 4°C, followed by purification via gel filtration (Superdex 200 396

    10/300 GL) in CSF-XB buffer. Approximately 70% labeling efficiency of the SNAP-tag was 397

    achieved. 398

    γ-tubulin was purified by binding to HisTrap HP (GE Healthcare) in binding buffer (50 399

    mM KPO4 pH 8.0, 500 mM KCl, 1 mM MgCl2, 10% glycerol, 5mM Imidazole, 0.25 µM GTP, 5 400

    mM BME, 2.5mM PMSF), washing first with 50 mM KPO4 pH 8.0, 300 mM KCl, 1 mM MgCl2, 401

    10% glycerol, 25 mM imidazole, 0.25 µM GTP, 5 mM BME), and then with 50 mM K-MES pH 402

    6.6, 500 mM KCl, 5mM MgCl2, 10% glycerol, 25 mM imidazole, 0.25 µM GTP, 5 mM BME) and 403

    eluted in 50 mM K-MES pH 6.6, 500 mM KCl, 5mM MgCl2, 10% glycerol, 250 mM imidazole, 404

    0.25 µM GTP, 5 mM BME. Peak fractions were further purified with gel filtration (Superdex 200 405

    10/300 GL) in buffer 50 mM K-MES pH 6.6, 500 mM KCl, 5 mM MgCl2, 1 mM K-EGTA, 1 µM 406

    GTP, 1 mM DTT. 407

    All proteins were flash-frozen and stored at -80°C, and their concentration was determined 408

    by analyzing a Coomassie-stained SDS-PAGE against known concentration of BSA (A7906, 409

    Sigma). 410

    411

    Purification, biotinylated and fluorescent labeling of γ-TuRC 412

    Endogenous γ-TuRC was purified from Xenopus egg extracts and labeled with the following steps 413

    at 4°C. 7-8 ml of meiotic extract from Xenopus laevis eggs, prepared as described previously45,46, 414

    was first diluted 5-fold with CSF-XBg buffer (10mM K-HEPES, 100mM KCl, 1mM MgCl2, 5mM 415

    K-EGTA, 10% w/v sucrose, 1mM DTT, 1mM GTP, 10 µg/ml LPC protease inhibitors, pH 7.7), 416

    centrifuged to remove large aggregates at 3500 rpm (Thermo Sorvall Legend XTR) for 10 minutes, 417

    and the supernatant filtered sequentially with 1.2 µm and 0.8 µm Cellulose Acetate filters 418

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 23

    (Whatman) followed by 0.22 µm PES filter (ThermoFisher). γ-TuRC was precipitated by 419

    incubating with 6.5% w/v PEG-8000k (Sigma) for 30 minutes and centrifuged at 17,000 rpm (SS-420

    34 rotor, ThermoScientific) for 20 minutes. γ-TuRC-rich pellet was resuspended in CSF-XB buffer 421

    with 0.05% v/v NP-40 using a mortar & pestel homogenizer, PEG was removed via centrifugation 422

    at 136,000 xg for 7 minutes in TLA100.3 (Beckman Ultracentrifuge), and supernatant was pre-423

    cleared by incubating with Protein A Sepharose beads (GE LifeSciences #17127901) for 20 424

    minutes. Beads were removed, γ-TuRC was incubated with 4-5 mg of a polyclonal antibody 425

    custom-made against C-terminal residues 413-451 of X. laevis γ-tubulin (Genscript) for 2 hours 426

    on gentle rotisserie, and further incubated with 1ml washed Protein A Sepharose bead slurry for 2 427

    hours. γ-TuRC-bound beads were washed sequentially with 30 ml of CSF-XBg buffer, 30 ml of 428

    CSF-XBg buffer with 250 mM KCl (high salt wash), 10 ml CSF-XBg buffer with 5mM ATP 429

    (removes heat-shock proteins), and finally 10 ml CSF-XBg buffer before labeling. For 430

    biotinylation of γ-TuRC, beads were incubated with 25 µM NHS-PEG4-biotin (A39259, 431

    ThermoFisher) in CSF-XBg buffer for 1 hour at 4°C, and unbound biotin was removed by washing 432

    with 30 ml CSF-XBg buffer prior to elution step. For combined fluorescent and biotin labeling of 433

    γ-TuRC, the wash step after ATP-wash consisted of 10 ml of labelling buffer (10mM K-HEPES, 434

    100mM KCl, 1mM MgCl2, 5mM K-EGTA, 10% w/v sucrose, 0.5mM TCEP, 1mM GTP, 10 µg/ml 435

    LPC, pH 7.2) and fluorescent labelling was performed by incubating the beads with 1 µM Alexa-436

    568 C5 Maleimide (A20341, ThermoFisher). Unreacted dye was removed with 10 ml CSF-XBg 437

    buffer, beads were incubated with 25 µM NHS-PEG4-biotin (A39259, ThermoFisher) in CSF-438

    XBg buffer for 1 hour at 4°C, and unreacted biotin removed with 30 ml CSF-XBg buffer. Labeled 439

    g-TuRC was eluted by incubating 2-3ml of g-tubulin peptide (residues 413-451) at 0.4mg/ml in 440

    CSF-XBg buffer with beads overnight. After 10-12 hours, g-TuRC was collected by adding 1-2ml 441

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 24

    CSF-XBg buffer to the column, concentrated to 200 µl in 30k NMWL Amicon concentrator (EMD 442

    Millipore) and layered onto a continuous 10-50 w/w % sucrose gradient prepared in a 2.2 ml ultra-443

    clear tube (11x34 mm, Beckman Coulter) using a two-step program in Gradient Master 108 444

    machine. Sucrose gradient fractionation of g-TuRC was performed by centrifugation at 200,000xg 445

    in TLS55 rotor (Beckman Coulter) for 3 hours. The gradient was fractionated from the top in 11-446

    12 fractions using wide-bore pipette tips and peak 2-3 fractions were identified by immunoblotting 447

    against g-tubulin with GTU-88 antibody (Sigma). g-TuRC was concentrated to 80 µl in 30k 448

    NMWL Amicon concentrator (EMD Millipore) and fresh purification was used immediately for 449

    single molecule assays. Cryo-preservation of g-TuRC molecules resulted in loss of ring assembly 450

    and activity. 451

    452

    Assessment of γ-TuRC with protein gel, immunoblot and negative stain electron microscopy 453

    To assess the purity of g-TuRC, 3-5 µl of purified g-TuRC was visualized on an SDS-PAGE with 454

    SYPRO Ruby stain (ThermoFisher) following the manufacturer’s protocol. Biotinylated subunits 455

    of g-TuRC were assessed by immunoblotting with Streptavidin-conjugated alkaline phosphatase 456

    (S921, ThermoFisher). g-TuRC purification was also assessed by visualizing using electron 457

    microscopy. 4 µl of peak sucrose gradient fraction of g-TuRC was pipetted onto CF400-Cu grids 458

    (Electron Microscopy Sciences), incubated at room temperature for 60 seconds and then wicked 459

    away. 2% uranyl acetate was applied to the grids for 30 seconds, wicked away, and the grids were 460

    air-dried for 10 minutes. The grids were imaged using Phillips CM100 TEM microscope at 64000x 461

    magnification. 462

    463

    Preparation of functionalized coverslips 464

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 25

    22x22 mm, high precision coverslips (170±5 µm, Carl Zeiss, catalog # 474030-9020-000) were 465

    functionalized for single molecule assays based on a recent protocol23,47 with specific 466

    modifications. Briefly, coverslips were labelled on the surface to be functionalized by scratching 467

    “C” on right, bottom corner, placed in Teflon racks, sonicated with 3N NaOH for 30 minutes, 468

    rinsed with water and sonicated in piranha solution (2 parts of 30 w/w % hydrogen peroxide and 469

    3 parts sulfuric acid) for 45 minutes. Coverslips were rinsed thrice in water, and all water was 470

    removed by spin drying completely in a custom-made spin coater. Pairs of coverslips were made 471

    to sandwich 3-glycidyloxypropyl trimethoxysilane (440167, Sigma) on the marked sides, placed 472

    in glass petri dishes, and covalent reaction was performed in a lab oven at 75°C for 30 minutes. 473

    Coverslips were incubated for 15 minutes at room temperature, the sandwiches were separated, 474

    incubated in acetone for 15 minutes, then transferred to fresh acetone and quickly dried under 475

    nitrogen stream. Coverslip sandwiches were prepared with a small pile of well mixed HO-PEG-476

    NH2 and 10% biotin-CONH-PEG-NH2 (Rapp Polymere) in glass petri dishes, warmed to 75°C in 477

    the lab oven until PEG melts, air bubbles were pressed out and PEG coupling was performed at 478

    75°C overnight. The following day, individual coverslips were separated from sandwiches, 479

    sonicated in MilliQ water for 30 minutes, washed further with water until no foaming is visible, 480

    dried with a spin dryer, and stored at 4°C. Functionalized coverslips were used within 1 month of 481

    preparation. 482

    Imaging chambers were prepared by first assembling a channel on glass slide with double 483

    sided tape strips (Tesa) 5 mm apart, coating the channel with 2mg/ml PLL(20)-g[3.5]- PEG(2) 484

    (SuSOS) in dH2O, incubating for 20 minutes, rinsing out the unbound PEG molecules with dH2O 485

    and drying the glass slide under the nitrogen stream. A piece of functionalized coverslip was cut 486

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 26

    with the diamond pen and assembled functionalized face down on imaging chamber. The prepared 487

    chambers were stored at 4°C and used within a day of assembly. 488

    489

    Microtubule nucleation assay with purified γ-TuRC, microscopy and data analysis 490

    The imaging channel was prepared as follows. First, 5% w/v Pluronic F-127 in dH2O was 491

    introduced in the chamber (1 vol = 50 µl) and incubated for 10 minutes at room temperature. The 492

    chamber was washed with 2 vols of assay buffer (80mM K-PIPES, 1mM MgCl2, 1mM EGTA, 493

    30mM KCl, 0.075% w/v methylcellulose 4000 cp, 1% w/v D-(+)-glucose, 0.02% w/v Brij-35, 494

    5mM BME, 1mM GTP) with 0.05 mg/ml κ-casien (casein buffer), followed by 1 vol of 0.5 mg/ml 495

    NeutrAvidin (A2666, ThermoFisher) in casein buffer, incubated on a cold block for 3 minutes, 496

    and washed with 2 vols of BRB80 (80mM K-PIPES, 1mM MgCl2, 1mM EGTA pH 6.8). 5-fold 497

    dilution of g-TuRC in BRB80 was introduced in the flow chamber and incubated for 10 minutes. 498

    Unattached g-TuRC molecules were washed with 1 vol of BRB80. 499

    During the incubations, nucleation mix was prepared containing desired concentration of 500

    αβ-tubulin (3.5-21 µM) purified from bovine brain with 5% Cy5-labeled tubulin along with 501

    1mg/ml BSA (A7906, Sigma) in assay buffer, centrifuged for 12 minutes in TLA100 (Beckman 502

    Coulter) to remove aggregates, a final 0.68 mg/ml glucose oxidase (SERVA, catalog # SE22778), 503

    0.16 mg/ml catalase (Sigma, catalog # SRE0041) was added, and reaction mixture was introduced 504

    into the flow chamber containing g-TuRC. 505

    506

    Total internal reflection fluorescence (TIRF) microscopy and analysis of microtubule 507

    nucleation from γ-TuRC 508

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 27

    Nucleation of MTs was visualized with inverted Nikon TiE TIRF microscope using a 100X, 1.49 509

    NA TIRF objective. An objective heater collar was attached (Bioptechs, model 150819-13) and 510

    the temperature set-point of 33.5°C was used for experiments. Time-lapse videos were recorded 511

    for 10 minutes at 0.5-1 frame per second using Andor iXon DU-897 camera with EM gain of 300 512

    and exposure time of 50-200 ms each frame. Reference time-point zero (0 seconds) refers to when 513

    the reaction was incubated at 33.5°C on the microscope, and for most reactions, imaging was 514

    started within 30 seconds. 515

    Growth speed of the plus-ends of MTs nucleated by g-TuRC was measured by generating 516

    kymographs in ImageJ. Region of interest (ROI) for individual MTs were selected and resliced to 517

    generate length-time plot, a line was fit to the growing MT, the slope of line represents growth 518

    speed. The kinetics of MT nucleation from g-TuRC was measured as follows. A kymograph was 519

    generated for every MT nucleated in the field of view. For most nucleation events, the time of 520

    nucleation of the MT was obtained from observing the kymograph and manually recording the 521

    initiation time point (see Fig. 1C for examples). For MTs where nucleation occurred before the 522

    timelapse movie began or where the initiation was not clearly observed in the kymograph, the 523

    shortest length of the MT that was clearly visible in the timelapse was measured and measured 524

    average growth speed of MTs was used to estimate the time of nucleation. We verified that this 525

    procedure accurately estimates the nucleation time for test case MTs where the nucleation event 526

    was visible. The measurement of number of MTs (N(t)) nucleated versus time was generated from 527

    a manual log containing the nucleation time for all MTs observed in the field of view, and a 528

    representative set of curves is displayed in Fig. 1F. A straight line was fit to the initial (linear) 529

    region of each N(t) versus t curve, rate of nucleation was obtained slope of each linear fit, and its 530

    power-law relation with tubulin concentration was obtained and reported (Fig. 1G). 531

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 28

    532

    Spontaneous microtubule nucleation and data analysis 533

    Spontaneous MT assembly was visualized similar to g-TuRC-mediated nucleation with the 534

    following changes. The pluronic, casein and NeutrAvidin incubations were performed identical to 535

    g-TuRC nucleation assay but instead of attaching g-TuRCs, sucrose-based buffer (of the same 536

    composition as used for g-TuRC elution) was diluted 5-fold with BRB80, introduced in the flow 537

    chamber and incubated for 10 minutes. Washes were performed with 1 vol of BRB80, nucleation 538

    mix was added, and imaging was performed as described above. MTs nucleate spontaneously in 539

    solution fall down on the coverslip due to depletion forces during the 10 minutes of visualizing the 540

    reaction. The number of MTs nucleated in the field of view were counted manually and plotted in 541

    Fig. 1I. 542

    543

    Preparation and microtubule assembly from blunt microtubule seeds 544

    Blunt MTs were prepared with GMPCPP nucleotide in two polymerization cycles as described 545

    recently22. Briefly, a 50 µl reaction mixture was prepared with 20 µM bovine brain tubulin with 546

    5% Alexa-568 labeled tubulin and 5% biotin-labeled tubulin, 1mM GMPCPP (Jena Bioscience) 547

    in BRB80 buffer, incubated on ice for 5 minutes, then incubated on 37°C for 30 minutes to 548

    polymerize MTs, and MTs were pelleted by centrifugation at 126,000 xg for 8 minutes at 30°C in 549

    TLA100 (Beckman Coulter). Supernatant was discarded, MTs were resuspended in 80% original 550

    volume of BRB80, incubated on ice for 20 minutes to depolymerize MTs, fresh GMPCPP was 551

    added to final 1mM, incubated on ice for 5 minutes, a second cycle of polymerization was 552

    performed by incubating the mixture at 37°C for 30 minutes, and MTs were pelleted again by 553

    centrifugation. Supernatant was discarded and MTs were resuspended in 200 µl warm BRB80, 554

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 29

    flash frozen in liquid nitrogen in 5µl aliquots, stored at -80°C and found to be stable for months. 555

    To verify that these MT seeds have blunt ends, frozen aliquots were quickly thawed at 37°C, 556

    diluted 20-fold with warm BRB80, and incubated at room temperature for 30 minutes to ensure 557

    blunt ends as described previously22. MTs were pipetted onto CF400-Cu grids (Electron 558

    Microscopy Sciences), incubated at room temperature for 60 seconds and then wicked away. 2% 559

    uranyl acetate was applied to the grids for 30 seconds, wicked away, and the grids were air-dried 560

    for 10 minutes. The grids were imaged using Phillips CM100 TEM microscope at 130000 x 561

    magnification and most MT ends were found to be blunt. 562

    To assay MT assembly from blunt MT seeds, MT assembly experiments similar to g-TuRC 563

    nucleation assays were performed with the following variation. A lower concentration 0.05 mg/ml 564

    NeutrAvidin (A2666, ThermoFisher) was attached, and washes were performed with warm 565

    BRB80 prior to attaching MTs. One aliquot of MT seeds was thawed quickly, diluted to 100-fold 566

    with warm BRB80, incubated in the chamber for 5 minutes, unattached seeds were washed with 1 567

    vol of warm BRB80, and the slide was incubated at room temperature for 30 minutes to ensure 568

    blunt MT ends. Wide bore pipette tips were used for handling MT seeds to minimize the shear 569

    forces that may result in breakage of MTs. Nucleation mix was prepared as described above and a 570

    low αβ-tubulin concentration (1.4-8.7 µM) was used. MT assembly from blunt seeds was observed 571

    immediately after incubating the slide on the objective heater. Imaging and analysis were 572

    performed as described above for to g-TuRC nucleation assays. However, the probability curves 573

    for MT assembly were obtained (Fig. 2B) by normalizing for the total number of seeds observed 574

    in the field of view. Rate of assembly was plotted against [tubulin concentration – C*], where C* 575

    represents the critical tubulin concentration below which MT ends do not polymerize obtained 576

    directly from experimental measurements (Fig. S2A-B). 577

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 30

    578

    Electron microscopy of γ-tubulin filaments in vitro 579

    Purified γ-tubulin was observed to form higher order oligomers previously using analytical gel 580

    filtration24. g-tubulin filaments were prepared by diluting pure g-tubulin to 1-5 µM to the buffer 581

    50mM K-MES pH 6.6, 5mM MgCl2, 1mM EGTA, 100mM KCl. g-tubulin mixture were pipetted 582

    onto CF400-Cu grids (Electron Microscopy Sciences), incubated at room temperature for 60 583

    seconds and then wicked away. 2% uranyl acetate was applied to the grids for 30 seconds, wicked 584

    away, and the grids were air-dried for 10 minutes. The grids were imaged using Phillips CM100 585

    TEM microscope at 130000 x magnification and g-tubulin filaments were seen to form. At 500 586

    mM KCl, g-tubulin filaments were not seen. 587

    588

    Nucleation of microtubules from purified γ-tubulin 589

    MT assembly experiments from purified g-tubulin was performed similar to g-TuRC nucleation 590

    assays described above with following variation. No avidin was attached to the coverslips, and 591

    varying concentration of g-tubulin was prepared by diluting purified g-tubulin in a high salt buffer 592

    (50mM K-MES pH 6.6, 500mM KCl, 5mM MgCl2, 1mM EGTA), centrifuging to remove 593

    aggregates separately for 12 minutes in TLA100 before adding to the nucleation mix containing 594

    15 µM αβ-tubulin (5% Cy5-labeled) with BSA, glucose oxidase and catalase as described above. 595

    The reaction mixture was introduced into the flow chamber and imaged via TIRF microscopy. A 596

    large number of MTs get nucleated immediately in the presence of 250 nM-1000 nM g-tubulin. 597

    598

    Measurement of affinity between purified γ-tubulin and αβ-tubulin 599

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 31

    Interaction assays between αβ-tubulin and g-tubulin were performed with biolayer interferometry 600

    using Octet RED96e (ForteBio) instrument in an 8-channel plate format. The plate temperature 601

    was held at 33°C and the protein samples were shaken at 400 rpm during the experiment. First, 602

    Streptavidin or anti-His antibody coated biosensors (ForteBio) were rinsed in interaction buffer 603

    (50mM K-MES pH 6.6, 100mM KCl, 5mM MgCl2, 1mM EGTA, 0.05% Tween20, 1mM GTP). 604

    100 nM biotin-labeled αβ-tubulin, or blank buffer, was bound to Streptavidin sensor, or 200 nM 605

    His-tagged g-tubulin to anti-His sensor until loaded protein results in a wavelength shift (Δλ) of 3 606

    nm. Unbound protein was removed by rinsing the sensor in interaction buffer, and interaction with 607

    αβ-tubulin was measured by incubating the sensor containing αβ-tubulin, g-tubulin or buffer with 608

    0-35 µM unlabeled αβ-tubulin in interaction buffer for 5 minutes. Δλ (nm) was recorded as a 609

    measure of the amount of unlabeled αβ-tubulin that binds to the sensor. Longitudinal interaction 610

    occurs between αβ-tubulin dimers and the resulting protofilaments were verified by visualizing 611

    the αβ-tubulin sample stained with 2% uranyl acetate using electron microscopy as described 612

    above (Fig. S2D). 613

    614

    Simulation of site occupation on γ-TuRC by αβ-tubulin dimers 615

    A simulation was performed in MATLAB for occupation of sites on g-TuRC by αβ-tubulin dimers. 616

    A circular grid was simulated with 13 empty positions that were occupied one per unit time 617

    stochastically such that a new position was selected by uniform random number generator and 618

    filled. If a previously filled position was selected, a different position was selected by the random 619

    number generator. The sequence in which the sites were occupied was followed. For each 620

    simulation, the total number of sites that were occupied when the first two neighboring sites are 621

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 32

    filled was recorded. The simulation was repeated 10,000 times and the probability of occurrence 622

    of first neighbor contact versus number of sites occupied is displayed in Fig. 2H. 623

    624

    Measuring the effect of microtubule associated proteins on γ-TuRC’s activity 625

    Effect of microtubule associated proteins (MAPs) was measured on g-TuRC’s nucleation activity. 626

    g-TuRC was attached on the coverslips using the setup described above and a control experiment 627

    was performed with identical reaction conditions for each protein tested. Nucleation mix was 628

    prepared containing 10.5 µM αβ-tubulin concentration (5% Cy5-labeled tubulin) as specified along 629

    with 1mg/ml BSA and oxygen scavengers, and either buffer (control), 10nM GFP-TPX2, 100nM 630

    EB1-mCherry, 5 µM Stathmin or 10nM MCAK was added. To test MCAK’s effect, the assay 631

    buffer additionally contained 1mM ATP. The reaction mixture containing tubulin and MAP at 632

    specified concentration was introduced into the flow chamber containing g-TuRC, and MT 633

    nucleation was visualized by imaging the Cy5-fluorescent channel at 0.5-1 frames per second. For 634

    TPX2 and EB1, fluorescence intensity of the protein was simultaneously acquired. The number of 635

    MTs nucleated over time was measured as described above and the effect of protein on g-TuRC’s 636

    nucleation activity was assessed by comparing nucleation curves with and without the MAP. 637

    A similar set of experiments were performed to study the effect of XMAP215 on g-TuRC-638

    mediated nucleation with the single molecule assays with the following differences. 20 nM of 639

    XMAP215-GFP was added to nucleation mix prepared with 3.5-7 µM αβ-tubulin concentration 640

    (5% Cy5-label) in XMAP assay buffer (80mM K-PIPES, 1mM MgCl2, 1mM EGTA, 30mM KCl, 641

    0.075% w/v methylcellulose 4000 cp, 1% w/v D-(+)-glucose, 0.007% w/v Brij-35, 5mM BME, 642

    1mM GTP). MTs nucleated from attached g-TuRC with and without XMAP215 were measured to 643

    assess the efficiency of nucleation induced by XMAP215 (Fig. 3C). To assess if C-terminal of 644

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 33

    XMAP215 increases nucleation efficiency, wild-type XMAP215 was replaced with a C-terminal 645

    construct of XMAP215: TOG5-Cterminus-GFP in the described experiment. 646

    To measure the kinetics of cooperative nucleation XMAP215 and g-TuRC, a constant 647

    density of g-TuRC was attached as described above and nucleation mix nucleation mix was 648

    prepared with a range of αβ-tubulin concentration between 1.6-7 µM (5% Cy5-label) with 20 nM 649

    of XMAP215-GFP in XMAP assay buffer, introduced into reaction chamber and MT nucleation 650

    was imaged immediately by capturing dual color images of XMAP215 and tubulin intensity at 0.5 651

    frames per second. 652

    653

    Triple-color imaging of XMAP215, γ-TuRC and microtubules 654

    For triple-color fluorescence assays (Fig. 3D), Alexa-568 and biotin-conjugated γ-TuRC was first 655

    attached to coverslips as described above with the following variation: 0.05 mg/ml of NeutrAvidin 656

    was used for attaching γ-TuRC. Nucleation mix was prepared with 7 µM αβ-tubulin (5% Cy5-657

    label), 10 nM Alexa-488 SNAP-tagged XMAP215 with BSA and oxygen scavengers in XMAP 658

    assay buffer (80mM K-PIPES, 1mM MgCl2, 1mM EGTA, 30mM KCl, 0.075% w/v 659

    methylcellulose 4000 cp, 1% w/v D-(+)-glucose, 0.007% w/v Brij-35, 5mM BME, 1mM GTP) 660

    and introduced into the reaction chamber containing attached γ-TuRC. Three-color imaging per 661

    frame was performed with sequential 488, 568 and 647 nm excitation and images were acquired 662

    with EMCCD camera at 0.3 frames per second. 663

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 34

    Figure legends 664

    665

    Figure 1. Microtubule nucleation by γ-TuRC. 666

    (A) Schematic of γ-TuRC mediated nucleation based on template model. (B) Purified, biotinylated 667

    γ-TuRC molecules were attached and time-lapse of MT nucleation is shown. Arrows point to 668

    nucleation sites. Representative kymographs of MTs nucleated from γ-TuRC are displayed in (C). 669

    The experiment and analyses in (B-G) were repeated at least thrice with independent γ-TuRC 670

    preparations. (D) Titrating tubulin concentration with constant the density of γ-TuRC. MT 671

    nucleation from γ-TuRC begins at 7 µM tubulin. (E) MT plus-end growth speed increases linearly 672

    with tubulin concentration. Linear fit (red line) with shaded 95% confidence intervals is displayed, 673

    with critical concentration for polymerization as C* = 1.4 µM. Inset: Number of MTs nucleated 674

    by γ-TuRCs within 120 seconds varies non-linearly with tubulin concentration. (F) Number of 675

    MTs nucleated (N(t)) over time (t) is plotted for varying tubulin concentration to obtain rate of 676

    nucleation as the slope of the initial part of the curves. (G) Number of tubulin dimers (n) in the 677

    critical nucleus on γ-TuRC was obtained as 3.7 ± 0.5 from the equation !"!# $#→& = ()#*+, 678

    displayed on a log-log plot. Data from two independent experiments was pooled and reported. (H) 679

    Spontaneous MT nucleation (schematized) was measured with increasing tubulin concentration 680

    and high concentrations. 14 µM tubulin is required. (I) Number of MTs (N(t=τ)) nucleated 681

    spontaneously were plotted against tubulin concentration (Ctub). Power-law curve was fit as N(t=τ) 682

    = k Ctubn and tubulin cooperativity (exponent) of n = 8 ± 1 was obtained. Experiments were 683

    repeated twice independently with many supporting results and all data were pooled. Scale bars, 684

    10 µm. See Figure S1 and Movies S1-S4. 685

    686

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 35

    Figure 2. Molecular mechanism for γ-TuRC-mediated microtubule nucleation. 687

    (A) Schematic and a micrograph of pre-formed, blunt MT seeds is shown and MT assembly from 688

    them was observed (right) with varying tubulin concentration. (B) Cumulative probability of MT 689

    assembly from seeds (p(t)) over time (t) is plotted and rate of nucleation was obtained as the slope 690

    from initial part of the curves. (C) Tubulin dimers (n) needed for MT assembly from seeds was 691

    from the relation !-!# $#→& = (()#*+ − )∗), displayed on a log-log plot. n = 1.2 ± 0.4 showing non-692

    cooperative assembly of tubulin. (D) MTs nucleate from purified γ-tubulin oligomers efficiently 693

    and (E) minus-ends of γ-tubulin-nucleated MTs remain capped while the plus-ends polymerize. 694

    (F) Molecular interaction between γ/αβ-tubulin was probed with bio-layer interferometry. Buffer 695

    (left), biotin-tagged αβ-tubulin (middle), or His-tagged γ-tubulin (right) were loaded on the probe 696

    as bait and untagged αβ-tubulin at 0-35 µM as prey. Wavelength shift, Δ3 (nm) indicated no 697

    binding between empty probe and αβ-tubulin or γ/αβ-tubulin, while that between αβ-/αβ-tubulin 698

    was observed and confirmed to be longitudinal (protofilament-wise, Fig. S2D). (G) Interface 699

    interaction model determines MT nucleation by γ-TuRC where lateral γ/γ-tubulin promote 700

    nucleation while low γ/αβ-tubulin affinity tunes nucleation. (H) Simulations were conducted where 701

    13 sites on γ-TuRC were stochastically occupied by αβ-tubulins. For two αβ-tubulin subunits to 702

    form lateral bond by occupying neighboring sites, 3.7 ± 1 subunits bind on average on γ-TuRC, 703

    predicting the size of critical nucleus. Experiments and analyses were repeated at least twice 704

    independently with multiple supporting results. Scale bars, 10 µm. See Figure S2 and Movie S5-705

    6. 706

    707

    Figure 3. Regulation of microtubule nucleation by TPX2 and XMAP215. 708

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 36

    (A) A constant density of γ-TuRC molecules were attached and 10.5µM tubulin ± 10nM GFP-709

    TPX2 were added. MTs were counted (right plot) and TPX2 was did not affect γ-TuRC-mediated 710

    nucleation. Scale bar, 10µm. (B) γ-TuRCs were attached and low concentration 3.5-7µM ± 20nM 711

    XMAP215 was added. XMAP215 induces MT nucleation from γ-TuRC efficiently. (C) MT 712

    nucleation events were counted and plotted. Scale bar, 10µm. (D) Sequence of events during 713

    cooperative MT nucleation by γ-TuRC and XMAP215 was visualized using labeled γ-TuRC 714

    (blue), XMAP215 (red) and tubulin (green). Time-lapse: γ-TuRC and XMAP215 form a complex 715

    prior to MT nucleation. XMAP215 variably resides on γ-TuRC for long (>100 seconds, 716

    kymograph 1) or short times (~3-10 seconds, kymograph 2) before MT nucleation and remains at 717

    the minus-end with 50% probability. Scale bar, 5µm. (E) Titrating tubulin with constant γ-TuRC 718

    and XMAP215 concentration. XMAP215/γ-TuRC nucleate MTs from 1.6 µM tubulin. Number of 719

    MTs nucleated (N(t)) over time (t) is plotted (inset) and rate of nucleation was obtained. Tubulin 720

    dimers (n) in critical nucleus was obtained as 3.2 ± 1.2 and displayed on a log-log plot. The 721

    experiment was performed once for all concentrations denoted and supported by a number of 722

    additional experiments. The remaining experiments were repeated more than twice with 723

    independent γ-TuRC preparations with additional supporting results. See Figure S3-4 and Movies 724

    S7-10. 725

    726

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 37

    Supplementary Figure legends 727

    728

    Supplementary Figure 1. Controls for γ-TuRC-mediated and spontaneous microtubule 729

    nucleation. 730

    (A-B) Protein gel (left) of purified γ-TuRC was stained with SYPRO Ruby stain and biotinylated 731

    sites on γ-TuRC visualized with alkaline phosphatase conjugated to avidin (right). Major, known 732

    γ-TuRC components were detected in the purified protein and GCP2/3 are heavily biotinylated 733

    during purification. Purified and biotinylated γ-TuRC was stained with uranyl acetate and 734

    visualized with transmission electron microscopy. Scale bar, 100nm. The experiments were 735

    repeated at least thrice with independent γ-TuRC preparations. 736

    (C) Covalent-reaction of biotin with γ-TuRC does not affect the nucleation activity, as measured 737

    by attaching γ-TuRC with anti-Mozart1 antibody and comparing the number of MTs nucleated by 738

    untagged and biotinylated γ-TuRC. Scale bar, 10µm. 739

    (D) Control reactions for γ-TuRC-mediated nucleation. MTs were nucleated by attaching purified 740

    γ-TuRC (left), adding control buffer (middle) or missing avidin in the reaction sequence (right). 741

    Robust MT nucleation only occurs with γ-TuRC attached to coverslips and not in control reactions. 742

    Scale bar, 10µm. See Movie S2. 743

    (E) MTs were first nucleated from γ-TuRC with Alexa 568-labeled tubulin (cyan), followed by 744

    introduction of Cy5-labeled tubulin (magenta). New tubulin only incorporates on the freely 745

    growing, plus-end but not at the nucleated minus-end. Scale bar, 10µm. The experiment was 746

    performed more than three times. 747

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 38

    (F) Two representative kymographs of spontaneously nucleated MTs are displayed, demonstrating 748

    that MTs grow from both the minus-end (dotted line) and the plus-end (solid line). Scale bar, 749

    10µm. See Movie S4. 750

    (G) MTs nucleation from γ-TuRCs or spontaneously were compared at two tubulin 751

    concentrations: 10.5 µM and 14 µM. γ-TuRC nucleates 10-fold higher number of MTs than 752

    spontaneous assembly. The experiment was performed twice with many supporting results. 753

    See also Figure 1. 754

    755

    Supplementary Figure 2. Microtubule assembly from blunt seeds and filament formation by 756

    purified γ-tubulin. 757

    (A) MT assembly (magenta) from MT seeds with blunt ends (cyan) was assayed. Tubulin 758

    concentration was titrated, and MT plus-end assembles starting from 2.45 µM tubulin, which is 759

    only slightly above the critical concentration of polymerization. Scale bar, 10µm. 760

    (B) Growth speed of MT plus-ends was measured from kymographs and critical concentration (C* 761

    = 1.4 µM) was determined from the linear fit (red line) with shaded 95% confidence intervals. The 762

    experiment and analyses in were repeated twice on independent days along with other supporting 763

    data. See also Figure 2 and Movie S5. 764

    (C) γ-tubulin self-assembles into filaments at high concentration and low-salt (100mM KCl) as 765

    imaged with negative-stain electron microscopy, whereas γ-tubulin filaments were not observed 766

    at high-salt (500mM KCl). Scale bar, 100nm. 767

    (D) Transmission electron microscopy of bio-layer interferometry assay of Fig. 2F show that 768

    protofilaments of αβ-tubulin form. The experiment was repeated twice. Scale bar, 100nm. 769

    770

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 39

    Supplementary Figure 3. Effect of microtubule associated proteins on γ-TuRC-mediated 771

    nucleation. 772

    (A) γ-TuRC molecules were attached to coverslips and either tubulin alone (pseudo-colored as 773

    magenta, left) or tubulin with 100nM EB1-mCherry (pseudo-colored as cyan, right) was added to 774

    the reaction. Number of MTs nucleated were measured (right plot) and EB1 was observed to 775

    neither increase nor decrease γ-TuRC-mediation nucleation despite functioning as a catastrophe 776

    factor in vitro. The experiment was repeated at least twice with independent γ-TuRC preparation. 777

    See also Movie S8. Scale bar, 10µm. 778

    (B) γ-TuRC molecules were attached to coverslips and either tubulin alone (left images), tubulin 779

    with 10nM MCAK (top right) or tubulin with 5µM Stathmin (bottom right) was added to the 780

    reaction. Both MCAK and Stathmin were observed to decrease the number of MTs nucleated 781

    because of their role in decreasing the net polymerization of a MT. The experiment was repeated 782

    at least twice with independent γ-TuRC preparations. Scale bar, 10µm. 783

    784

    Supplementary Figure 4. Cooperative microtubule nucleation XMAP215 and γ-TuRC. 785

    (A) γ-TuRC molecules were attached and increasing concentration of tubulin was added with 786

    20nM XMAP215. XMAP215 was found to induce MT nucleation from γ-TuRC molecules at even 787

    low tubulin concentration of 1.6-3.5 µM where γ-TuRCs alone do not nucleate MTs. See Figure 788

    3E. Scale bar, 10µm. 789

    (B) The role of C-terminal region of XMAP215 was tested in cooperative nucleation with γ-TuRC. 790

    MTs nucleated by γ-TuRC alone (left), γ-TuRC with 20nM full-length XMAP215 (middle) or γ-791

    TuRC with 20nM C-terminal domain of XMAP215 were visualized. The C-terminal domains of 792

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 40

    XMAP215 do not stimulate MT nucleation from γ-TuRC. The experiment was repeated twice with 793

    independent γ-TuRC preparations. Scale bar, 10µm. 794

    795

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 41

    Movie Legends 796

    797

    Movie 1. Microtubule nucleation from γ-TuRC complexes 798

    γ-TuRC was attached to functionalized coverslips and MT nucleation was observed upon 799

    introducing fluorescent αβ-tubulin (gray). MTs nucleated from individual γ-TuRC molecules from 800

    zero length at 15µM αβ-tubulin and the plus-end of nucleated MTs polymerized, but not its minus-801

    end. Elapsed time is shown in seconds, where time-point zero represents the start of reaction. Scale 802

    bar, 10 µm. 803

    804

    Movie 2. Microtubule nucleation from γ-TuRC is specific 805

    γ-TuRC was immobilized on coverslips (leftmost panel) and MT nucleation was observed upon 806

    introducing fluorescent αβ-tubulin (gray). Control reactions where either no γ-TuRC was added 807

    (middle panel) or γ-TuRC was not specifically attached (rightmost panel) did not result in MT 808

    nucleation. Elapsed time is shown in seconds, where time-point zero represents the start of 809

    reaction. Scale bar, 10 µm. 810

    811

    Movie 3. γ-TuRC molecules nucleated microtubules efficiently 812

    Constant density of γ-TuRC was attached while concentration of fluorescent αβ-tubulin was 813

    titrated (3.5-21µM) and MT nucleation was observed. γ-TuRC molecules nucleated MTs starting 814

    from 7µM tubulin and MT nucleation increased non-linearly with increasing tubulin concentration. 815

    Elapsed time is shown in seconds, where time-point zero represents the start of reaction. Scale bar, 816

    10 µm. 817

    818

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 42

    Movie 4. Spontaneous microtubule nucleation occurs at high tubulin concentration 819

    Concentration of fluorescent αβ-tubulin was titrated (7-21µM) and spontaneous MT nucleation 820

    was assayed. MTs nucleated spontaneously starting from high concentration of 14µM tubulin and 821

    MT nucleation increased non-linearly with tubulin concentration. Both plus- and minus-ends of 822

    the assembled MTs polymerize. Elapsed time is shown in seconds, where time-point zero 823

    represents the start of reaction. Scale bar, 10 µm. 824

    825

    Movie 5. Microtubule assembly from blunt plus-ends resembles polymerization 826

    MTs with blunt ends (seeds, cyan) were generated and attached to functionalized coverslips. 827

    Varying concentration of fluorescent αβ-tubulin was added (1.4-8.7µM, pseudo-colored as 828

    magenta) and MT assembly from seeds was assayed. MTs assembled at concentration above 829

    1.4µM tubulin, which is the minimum concentration needed for polymerization of MT plus-ends 830

    (C*). MT assembly from seeds increased linearly with the concentration of assembly-competent 831

    tubulin (C-C*). Elapsed time is shown in seconds, where time-point zero represents the start of 832

    reaction. Scale bar, 10 µm. 833

    834

    Movie 6. Arrays of purified γ-tubulin nucleate microtubules 835

    Purified γ-tubulin nucleated MTs. Fluorescent αβ-tubulin (10.5µM, colored as gray) was added to 836

    purified γ-tubulin at increasing concentration, and MT nucleation was assessed. MTs assembled 837

    from 250-1000 nM γ-tubulin, where γ-tubulin alone self-assembled into higher order oligomers 838

    and filaments in lateral γ/γ-tubulin arrays. Minus-ends of γ-tubulin-nucleated MTs did not 839

    polymerize, while the plus-ends did. Elapsed time is shown in seconds, where time-point zero 840

    represents the start of reaction. Scale bar, 10 µm. 841

    not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which wasthis version posted November 23, 2019. ; https://doi.org/10.1101/853010doi: bioRxiv preprint

    https://doi.org/10.1101/853010

  • 43

    842

    Movie 7. TPX2 does not increase γ-TuRC’s microtubule nucleation activity 843

    γ-TuRC was immobilized on coverslips and MT nucleation was observed upon introducing 844

    fluorescent αβ-tubulin (10.5µM, pseudo-colored as magenta) without or with 10nM GFP-TPX2 845

    (pseudo-colored as cyan) in the left and right panels respectively. TPX2 bound along the nucleated 846

    MTs but did not increase the MT nucleation activity of γ-TuRC molecules. Elapsed time is shown 847

    in seconds, where time-point zero represents the start of reaction. Scale bar, 10 µm. 848

    849

    Movie 8. EB1 does not decrease the microtubule nucleation activity of γ-TuRC 850

    γ-TuRC was immobilized on coverslips and MT nucleation was observed upon introducing 851

    fluorescent αβ-tubulin (10.5µM, pseudo-colored as magenta) without or with 100nM EB1-852

    mCherry (pseudo-colored as cyan) in the left and right panels respectively. EB1 binds the plus-853

    ends of nucleated MTs but did not decrease the MT nucleation activity of γ-TuRC molecules. 854

    Elapsed time is shown in seconds, where time-point zero represents the start of reaction. Scale bar, 855

    10 µm. 856

    857

    Movie 9. XMAP215 increases microtubule nucleation activity of γ-TuRC 858

    γ-TuRC was immobilized on coverslips and MT nucleation was assayed with low concentratio